Allelotype analysis of adult T-cell leukemia (ATL) was undertaken for the first time to identify chromosomal loci relevant to the development of acute/lymphomatous ATL. Loss of heterozygosity (LOH) was screened using 94 highly polymorphic microsatellite markers, distributed among all nonacrocentric, autosomal chromosomes. In each of the 22 cases, DNA obtained from their leukemic cells in acute/lymphomatous phase was compared with their constitutional DNA from mononuclear cells in chronic or remission phase. Allelic losses of at least on one chromosome arm occurred in 91% of the cases (20 individuals). Among 39 chromosome arms, allelic losses were observed on 31 arms at least for one sample. A high frequency of allelic loss (>30%) was seen on chromosome arms 6q (41%) and 17p (48%). The mean fractional allelic loss (FAL) was 0.109. These findings suggest that a novel tumor suppressor gene on chromosome arm 6q, as well as the p53 gene on chromosome arm 17p, probably have an important role in the development of acute/lymphomatous ATL.

© 1998 by The American Society of Hematology.

INITIATION AND PROGRESSION of neoplasia can include inactivation of one or several tumor suppressor genes. The paradigm of alterations of a tumor suppressor gene is a mutation of one allele and loss of the second allele.1 Such losses may involve deletions too small to be detected by conventional cytogenetics. This reduction to homozygosity can be detected by the finding of loss of heterozygosity (LOH) of polymorphic sequences in tumor DNA as compared with constitutional DNA from the same individual. Several tumor suppressor genes, such as the DCC gene, have been cloned from the hot spot of LOH analysis.2-5 Thus, LOH analysis is an indirect method to search for a tumor suppressor gene. Allelotype analysis is an extensive survey of allelic loss throughout the genome, screening multiple loci for regions containing tumor suppressor genes that are likely to be altered.5-15 This technique has recently become extremely powerful with the use of microsatellite markers, which are short tracts of (CA)n repeats with high polymorphism that exists throughout the genome.16,17 

Adult T-cell leukemia (ATL) is an aggressive, fatal malignancy of mature CD4+ T lymphocytes.18,19 The human T- cell lymphotropic virus type I (HTLV-I) has been recognized as the etiologic agent of ATL.20-22 However, a long period of clinical latency (mean, 55 years) precedes the developement of ATL23 and only a small percentage of HTLV-I-infected individuals develop this malignancy (one patient/1,000 to 2,000 carriers/year),24,25 indicating that additional genetic events probably are required to develop ATL after viral infection of the target T cells. Statistical analysis suggests that ATL arises following five independent genetic events.26 

Studies to date by ourselves and others have implicated deletions or mutations of several tumor suppressor genes in the pathogeneisis of ATL, including p53 in about 40% of acute/lymphomatous ATL,26-28 and the p15INK4B, p16INK4A, and Rb genes in approximately 30%, 35%, and 5% of such cases, respectively.29-32 In addition, loss of expression of p18INK4C was found in ATL cell lines.33 To begin to define further genetic events leading to the evolution of acute/lymphomatous ATL, we performed comprehensive allelotype analysis of ATL using 94 microsatellite markers that covered all autosomal chromosomes.

Samples.

Patients with acute/lymphomatous ATL were investigated; 21 samples were from Japanese patients and one was from an individual from Panama. The percentage of contaminating normal cells in the acute/lymphomatous phase samples was at most 30% and usually less than 10%. The corresponding control DNAs were obtained from either their peripheral blood after complete remission (n = 17) or during their chronic phase (n = 5). The clinical subtypes of ATL were based on the diagnostic criteria proposed by the Lymphoma Study Group of Japan.34Cytogenetic data were available from five patients. All five cases showed chromosomal abnormalities: patient O, 45X,add(X),-4, -6,del(7)(p15),add(10)(p11), add(11)(q23), add(12)(q11),add(14)(p11),- 21, add(22)(p11),+2mar[6]/46XY[7]; patient P, 45XY,-3; patient Q, 45X,-Y,add(3)(p11q21), add(7)(p11), del(9)(q13q21),add(10)(p13),add(12)(q13), der(15)t(3;15)(p14;p11),-20,+mar[14]/46, idem+Y[10]/46XY[10]; patinet S, 48XY,+2,-3,-6, +add(6)(q32),-10,+add(7)(p13), add(7)(q32),-10, +add(10)(q22), +13,-15,-18,-21,+6,mar; patient U, 47XX,+mar[1]/46XX[3].

Allelic loss analysis.

Primers for polymerase chain reaction (PCR) amplification of microsatellite markers, chosen to represent at least two loci on each chromosomal arm, were obtained from Research Genetics (Huntsville, AL). Ninty-four markers distributed among all 39 autosomal chromosome arms, except for nonacrocentric arms, were analyzed. These markers and their chromosomal regions are detailed in Table1. PCRs were performed as described.35 Briefly, total reaction volumes were 20 μL containing 25 ng DNA, 1.5 mmol/L MgCl2, 10 pmol/L of each of the primers, 2 nmol/L of each of the four deoxyribonucleotide triphosphates (dNTP; Pharmacia, Stockholm, Sweden), 1 U of Taq DNA polymerase (GIBCO-BRL, Gaithersburg, MD), and 2 μCi 32P-labeled deoxycytidine triphosphates (dCTP) (3,000 μCi/mmol; New England Nuclear/Dupont, Boston, MA) with specified buffer provided by the supplier. To ascertain either LOH or duplication of the region, PCR reaction was performed in a multiplex fashion for some of the markers; the reaction mixture contained two primer sets. PCR consisted of 40 seconds at 94°C, 30 seconds at 55°C, and 1 minute at 72°C for 27 to 33 cycles in a Programmable Thermal Controller (MJ Research Inc, Water Town, MA) in order to examine the products in the linear range of signals. PCR products were mixed with a formamide gel-loading solution, heat denatured at 94°C, separated on a denaturing 5% to 8% polyacrylamide gel, and visualized by autoradiography. Allele losses were determined by visual inspection. When visible reduction of radiographic signal was equivocal, a radioanalytic imaging detector (Ambis, Ambis Inc, San Diego, CA) was used to confirm our interpretation. All positive results were repeated for confirmation.

Table 1.

Microsatellite Markers Used for Each Chromosomal Arm for Allelotyping of ATL

Chromosome Arm Markers
1p  D1S199, D1S244  
1q D1S196, D1S251  
2p  D2S113, D2S123, D2S146  
2q  D2S122, D2S155  
3p  D3S1266, D3S1285  
3q  D3S1272, D3S1299  
4p D4S418, D4S419  
4q  D4S408, D4S416  
5p  D5S416, D5S418 
5q  APC, MDF27  
6p  D6S260, D6S265  
6q  D6S284, D6S294, D6S1601, D6S1627  
7p  D7S493, D7S510, D7S517  
7q D7S487, D7S522, D7S677  
8p  D8S262, D8S283  
8q  D8S272, D8S286  
9p  D9S126, D9S157, D9S165, D9S168, D9S171, INFA  
9q D9S154, D9S176  
10p  D10S191, D10S197  
10q  D10S190, D10S201  
11p  D11S902, D11S907  
11q  D11S924, D11S1391, D11S2179  
12p  D12S89, D12S308, D12S364  
12q  D12S84, D12S346  
13q  D13S156, D13S221  
14q  D14S61, D14S64, D14S72 
15q  D15S126, D15S128, D15S165  
16p  D16S406, D16S418 
16q  D16S402, D16S514  
17p  MDF41, TP53, D17S786  
17q D17S802, D17S805  
18p  D18S54, D18S452  
18q  DCC, D18S58, D18S67  
19p  D19S221, D19S226, D19S424  
19q D19S180, D19S214  
20p  D20S98, D20S116  
20q  D20S100, D20S108  
21q  D21S265, D21S270  
22q  D22S280, D22S283 
Chromosome Arm Markers
1p  D1S199, D1S244  
1q D1S196, D1S251  
2p  D2S113, D2S123, D2S146  
2q  D2S122, D2S155  
3p  D3S1266, D3S1285  
3q  D3S1272, D3S1299  
4p D4S418, D4S419  
4q  D4S408, D4S416  
5p  D5S416, D5S418 
5q  APC, MDF27  
6p  D6S260, D6S265  
6q  D6S284, D6S294, D6S1601, D6S1627  
7p  D7S493, D7S510, D7S517  
7q D7S487, D7S522, D7S677  
8p  D8S262, D8S283  
8q  D8S272, D8S286  
9p  D9S126, D9S157, D9S165, D9S168, D9S171, INFA  
9q D9S154, D9S176  
10p  D10S191, D10S197  
10q  D10S190, D10S201  
11p  D11S902, D11S907  
11q  D11S924, D11S1391, D11S2179  
12p  D12S89, D12S308, D12S364  
12q  D12S84, D12S346  
13q  D13S156, D13S221  
14q  D14S61, D14S64, D14S72 
15q  D15S126, D15S128, D15S165  
16p  D16S406, D16S418 
16q  D16S402, D16S514  
17p  MDF41, TP53, D17S786  
17q D17S802, D17S805  
18p  D18S54, D18S452  
18q  DCC, D18S58, D18S67  
19p  D19S221, D19S226, D19S424  
19q D19S180, D19S214  
20p  D20S98, D20S116  
20q  D20S100, D20S108  
21q  D21S265, D21S270  
22q  D22S280, D22S283 

A panel of 94 highly informative microsatellite markers representing every autosomal chromosome were used to screen 22 matched ATL samples. Although PCR allelotyping is capable of showing losses of genetic material in tumors, it is unsuitable for detection of gene amplifications. To rule out the possibility of gene amplification, we performed multiplex PCR to compare the intensity of two loci. Because gene amplification was not found, each of the allelic imbalances were determined to be LOH. Representative examples of autoradiograms demonstrating LOH are shown in Fig 1.

Fig. 1.

Autoradiographs from LOH analysis with microsatellite markers. PCR products were separated by polyacrylamide gel electrophoresis, showing examples of allelic loss found in patient E. DNA samples isolated from leukemic cells in the acute phase are labelled as A and those DNA samples isolated from corresponding normal peripheral leukocytes after complete remission are designated as C. Microsatellite markers are indicated above the autoradiograms. Arrows, loss of one allele.

Fig. 1.

Autoradiographs from LOH analysis with microsatellite markers. PCR products were separated by polyacrylamide gel electrophoresis, showing examples of allelic loss found in patient E. DNA samples isolated from leukemic cells in the acute phase are labelled as A and those DNA samples isolated from corresponding normal peripheral leukocytes after complete remission are designated as C. Microsatellite markers are indicated above the autoradiograms. Arrows, loss of one allele.

Close modal

For each patient, we obtained information for at least 30 (76.9%) chromosomal arms (Table 2). All but two ATLs (patients N and U) showed evidence of a deletion at one or more loci with loss(es) occurring in one to 13 chromosomal arms. Sixteen samples showed losses in more than two different chromosomal arms. Patients E and G had allele loss affecting over 30% of informative chromosomal arms.

Table 2.

Allelic Losses in Chromosomal Arms and FAL

Samples Chromosomal Arms With LOH No. of Informative Arms FAL
A  16p,  30  0.033  
B  20p, 20q,  31  0.065  
C  2p, 7q, 8p, 17q, 18q, 22q,  35 0.171  
D  3q, 4p, 5q, 6q, 9p, 18q, 22q,  34  0.206 
E  2q, 6p, 6q, 9p, 9q, 14q, 17p, 17q, 18q, 22q, 30 0.333  
F  6q, 17p, 18q,  34 0.088  
G  6p, 6q, 8p, 10p, 12p, 12q, 13q, 16p, 17p, 17q, 18q, 19p, 22q,  35 0.371  
H  6p, 6q, 7q,  32 0.094  
I  10q,  33  0.030  
J  17p,  31 0.032  
K  17p,  34  0.029  
L  6q, 4q, 13q, 17p, 37  0.108  
M  6p, 17q,  33  0.061  
N   31 0  
O  2q, 4p, 4q, 12p, 12q, 17p,  36  0.167  
6q, 7p, 7q, 12p,  36  0.111  
Q  8q, 17p,  31 0.065  
R  7p,  37  0.027  
S  4p, 6q, 7p, 7q, 8p, 10p, 10q, 18p,  34  0.235  
T  6q, 2p, 20q,  33  0.091 
U   33  0  
V  7q, 9p, 17p,  33  0.091 
Samples Chromosomal Arms With LOH No. of Informative Arms FAL
A  16p,  30  0.033  
B  20p, 20q,  31  0.065  
C  2p, 7q, 8p, 17q, 18q, 22q,  35 0.171  
D  3q, 4p, 5q, 6q, 9p, 18q, 22q,  34  0.206 
E  2q, 6p, 6q, 9p, 9q, 14q, 17p, 17q, 18q, 22q, 30 0.333  
F  6q, 17p, 18q,  34 0.088  
G  6p, 6q, 8p, 10p, 12p, 12q, 13q, 16p, 17p, 17q, 18q, 19p, 22q,  35 0.371  
H  6p, 6q, 7q,  32 0.094  
I  10q,  33  0.030  
J  17p,  31 0.032  
K  17p,  34  0.029  
L  6q, 4q, 13q, 17p, 37  0.108  
M  6p, 17q,  33  0.061  
N   31 0  
O  2q, 4p, 4q, 12p, 12q, 17p,  36  0.167  
6q, 7p, 7q, 12p,  36  0.111  
Q  8q, 17p,  31 0.065  
R  7p,  37  0.027  
S  4p, 6q, 7p, 7q, 8p, 10p, 10q, 18p,  34  0.235  
T  6q, 2p, 20q,  33  0.091 
U   33  0  
V  7q, 9p, 17p,  33  0.091 

Abbreviation: FAL, fractional allelic loss.

Of the 39 chromosomal arms anaylzed, 29 (74.4%) showed LOH in one or more samples. Figure 2 summarizes the percentage of allelic loss at each chromosomal arm, which was calculated by dividing the number of tumors with LOH at any marker on the chromosmome arm by the total number of informative cases. Frequency of LOH for individual chromosomal arms varied between 0% (1p, 1q, 3p, 5p, 11p, 11q, 15q, 16q, 19q, and 21q) and 47.6% (17p). Over 30% of informative acute/lymphomatous samples showed LOH at chromosomal arms 6q (9/22, 40.9%) and 17p (10/21, 47.6%). Of the 22 samples, five, six, and four cases had LOH on 6q alone, 17p alone, and both 6q and 17p, respectively. In total, 15 samples had LOH on 6q and/or 17p.

Fig. 2.

Frequency of allelic loss on each chromosome arm in 22 acute/lymphomatous ATLs. Percentage of LOH was calculated for each chromosome arm by dividing the number of tumors with LOH at any marker on the chromosmome arm by the total number of informative cases. (□) p; (▪) q.

Fig. 2.

Frequency of allelic loss on each chromosome arm in 22 acute/lymphomatous ATLs. Percentage of LOH was calculated for each chromosome arm by dividing the number of tumors with LOH at any marker on the chromosmome arm by the total number of informative cases. (□) p; (▪) q.

Close modal

Fractional allelic loss (FAL) was calculated as the ratio of chromosomal arms that showed loss among all informative arms for each patient, as described by Vogelstein.2 The FAL values in our samples varied from 0 to 0.371, with a mean of 0.109 (Table 2).

To our knowledge, this is the first genomic search in ATL using microsatellite markers; the aim was to identify chromosomal regions likely to contain important tumor suppressor genes involved in the progression of acute/lymphomatous ATL. The analysis showed two hot-spots of LOH (>30%), one on 6q and the other one on 17p. Although it is possible that candidate tumor suppressor genes are present in the undeleted segments of DNA, our result suggests that tumor suppressor genes for progression of acute/lymphomatous ATL probably reside on chromosome 6q and 17p.

With regard to tumor suppressor genes, mutations of p53 are among the most common genetic changes in human tumors.36 The greatest frequency of LOH in acute/lymphomatous ATL was observed at the TP53 locus (seven of 15, 46.7%) on chromosome 17p. This result is congruent with previous reports by ourselves and others that the p53 gene was mutated in about 40% of acute/lymphomatous ATL samples.26-28 Although point mutations of the p53 gene in our samples were not examined, we suspect that those with LOH at TP53 had mutations of the gene.

The high frequency of allelic loss affecting 6q is a novel finding in ATL and suggests that this is the site of an as yet uncharacterized tumor suppressor gene(s). The LOH for 6q is in keeping with results in several other types of tumors, including those of the kidney,37 breast,4,38 ovary,39,40liver,41 and prostate,42 as well as melanoma,43-46 lymphoma/lymphoblastic leukemia,47-52 and parathyroid adenoma.53Further studies are required to define the altered gene on chromosome 6q in ATL and determine whether inactivation of the same gene occurrs in all of the cancer types with LOH in this chromosomal region.

The tumor suppressor gene p16INK4A was mapped to chromosome 9p21.54,55 We previously found that this gene was deleted in about 35% of acute/lymphomatous ATL samples.28,29 In this study, we analyzed for LOH at D9S171, which maps to 9p21 about 1 megabase proximal to the p16INK4A gene. Only eight cases were informative at D9S171 and two of them had LOH. The frequency of heterozygosity for this locus is lower than that in the Genome Database (0.79). Such discordance could be due to homogeneity of the Japanese population. In addition, we may have missed some homozygous deletions because homozygous deletions for a polymorphic marker may appear as retention of heterozygosity as a consequence of amplification of DNA from the contaminating normal cells. Therefore, we could have underestimated the frequency of LOH at D9S171.

ATL is frequently characterized by an acquisition of nonrandom chromosomal changes,56 but cytogenetic changes from chronic to acute ATL have been rarely reported. Additionally, small deletions are below the limits of resolution of cytogenetic analysis. Karyotype analyses were available for only five samples in our study. As expected, in four of these five samples, LOH studies identified additional abnormalities not detected in the cytogenetic analysis. Thus, cytogenetic studies have probably missed most of the interstitial deletions. Nevertheless, some of the chromosomal deletions shown by cytogenetic analyses were not detected by our LOH analyses. Most of these samples had noninformative microsatellite loci in these regions. The discrepancy between cytogenetic and LOH studies in several samples can be ascribed to a failure to examine a microsatellite marker in the deleted chromosomal region.

The mean FAL was 0.109 in acute/lymphomatous ATL, which is lower than that in solid tumors.4,7,10,57,58 This suggests that ATL cells may be genetically more stable than those of solid tumor cells.

In summary, the comprehensive allelotyping of ATL has provided novel signposts for future efforts at positional candidate cloning of human tumor suppressor genes involved in progression of ATL. Likewise, these genes will be likely targets for development of other hematologic malignancies, as well as other human tumors.

Supported in part by Grants No. CA26038, CA42710, and CA70675 from the National Institues of Health, Bethesda, MD; the Concern Foundation, Beverly Hills, CA; and the Parker Hughes Fund, Glendale, CA.

Address reprint requests to Yoshihiro Hatta, MD, Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Research Institute, UCLA School of Medicine, 8700 Beverly Blvd, B-208, Los Angeles, CA 90048.

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

1
Knudson
AG
Mutation and cancer. Statistical study of retinoblastoma.
Proc Natl Acad Sci USA
68
1971
820
2
Fearon
ER
Cho
KR
Nigro
JM
Kern
SE
Simons
JW
Ruppert
JM
Hamilton
SR
Preosomger
AC
Thomas
G
Kinzler
KW
Vogelstein
B
Identificaion of a chromosome 18q gene that is altered in colorectal cancers.
Science
247
1990
49
3
Steck
PA
Pershouse
MA
Jasser
SA
Yung
WKA
Lin
H
Ligon
AH
Langford
LA
Baumgard
ML
Hattier
T
Davis
T
Frye
C
Hu
R
Swedlund
B
Teng
DHF
Tavtigian
SV
Identificaion of a candidate tumor suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers.
Nat Genet
15
1997
356
4
Theile
M
Seitz
S
Arnold
W
Jandrig
B
Frege
R
Schlag
PM
Haensch
W
Winzer
K-J
Barett
JC
Scherneck
S
A defined chromosome 6q fragment (at D6S310) harbors a putative tumor supressor gene for breast cancer.
Oncogene
13
1996
677
5
Vogelstein
B
Fearon
ER
Kern
SE
Hamilton
SR
Preisinger
AC
Nakamura
Y
White
R
Allelotype of colorectal carcinomas.
Science
244
1989
207
6
Cliby
W
Ritland
S
Hartmann
L
Dodson
M
Halling
KC
Keeney
G
Podratz
KC
Jenkins
RB
Human epithelial ovarian cancer allelotype.
Cancer Res
53
1993
2393
7
Nawroz
H
van der Riet
P
Hruban
RH
Koch
W
Ruppert
JM
Sidransky
D
Allelotype of head and neck squamous cell carcinoma.
Cancer Res
54
1994
1152
8
Knowles
MA
Elder
PA
Willamson
M
Cairns
JP
Shaw
ME
Law
MG
Allelotype of human bladder cancer.
Cancer Res
54
1994
531
9
Mitra
AB
Murty
VVVS
Li
RG
Pratrap
M
Luthra
UK
Chaganti
RSK
Allelotype analysis of cervical carcinoma.
Cancer Res
54
1994
4481
10
Fujino
T
Risinger
JI
Cliins
NK
Liu
F-S
Nishii
H
Takahashi
H
Westphal
E-M
Barett
JC
Sasaki
H
Kohler
MF
Berchuck
A
Boyd
J
Allelotype of endometrial carcinoma.
Cancer Res
54
1994
4294
11
Seymour
AB
Hruban
RH
Redston
M
Caldas
C
Powell
SM
Kinzler
KW
Yeo
CJ
Kern
SE
Allelotype of pancreatic adenocarcinoma.
Cancer Res
54
1994
2761
12
Osborn
RJ
Leech
V
Polymerase chain reaction allelotyping of human ovarian cancer.
Br J Cancer
69
1994
429
13
Takeuchi
S
Bartram
CR
Wada
M
Reiter
A
Hatta
Y
Seriu
T
Lee
E
Miller
CW
Miyoshi
I
Koeffler
HP
Allelotype analysis of childhood acute lymphoblastic leukemia.
Cancer Res
15
1995
5377
14
Pabst
T
Schwaller
J
Bellomo
MJ
Oestreicher
M
Mühlematter
D
Tichelli
A
Tobler
A
Fey
MF
Frequent clonal loss of heterozygosity but scarcity of microsatellite instability at chromosomal breakpoint cluster regions in adult leukemia.
Blood
88
1996
1026
15
Mori
N
Morosetti
R
Lee
S
Spira
S
Ben-Yahuda
D
Schiller
G
Landolfi
R
Mizoguchi
H
Koeffler
HP
Allelotype analysis in the evolution of chronic myelocytic leukemia.
Blood
90
1997
2010
16
Litt
M
Luty
JA
A hypervariable microsatellite revealed by in vitro amplification of a dinucleotide repeat within the cardiac muscle actin gene.
Am J Hum Genet
44
1989
397
17
Weber
JL
May
PE
Abundant class of human DNA polymorphisms which can be typed using the polymerase chain reaction.
Am J Hum Genet
44
1989
388
18
Uchiyama
T
Yodoi
J
Sagawa
K
Takatsuki
K
Uchino
H
Adult T-cell leukemia: Clinical and hematologic features of 16 cases.
Blood
50
1977
481
19
Hattori
T
Uchiyama
T
Tobinai
T
Takatsuki
T
Uchino
H
Surface phenotype of Japanese ATL cells characterized by MoAbs.
Blood
58
1981
645
20
Miyoshi
I
Kubonishi
I
Yoshimoto
S
Akagi
T
Ohtsuki
Y
Shiraishi
Y
Nagata
K
Hinuma
Y
Type C virus particles in a cord T-cell line derived by co-cultivating normal human cord leukocytes and human leukemic T cells.
Nature
294
1981
770
21
Yoshida
M
Miyoshi
I
Hinuma
Y
Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease.
Proc Natl Acad Sci USA
79
1982
2031
22
Popovic
M
Sarin
PS
Robert-Guroff
M
Kalyanraman
VS
Mann
D
Minowada
J
Gallo
RC
Isolatioin and transmission of human retrovirus (human T-cell leukemia virus).
Science
219
1983
856
23
Tajima
K
The T- and B-cell malignancy Study Group
The fourth nationwide study of adult T-cell leukemia/lymphoma (ATL in Japan): Estimates of the risk of ATL and its geographical and clinical features.
Int J Cancer
45
1990
237
24
Tokudome
S
Tokunaga
O
Shimamoto
Y
Miyamoto
Y
Sumida
I
Kikuchi
M
Takeshita
M
Ikeda
T
Fujiwara
K
Yoshihara
M
Yanagawa
T
Nishizumi
M
Incidence of adult T-cell leukemia/lymphoma among human T-lymphotropic virus type 1 carriers in Saga, Japan.
Cancer Res
49
1989
226
25
Okamoto
T
Multi-step carcinogenesis model for adult T-cell leukemia.
Jpn J Cancer Res
80
1989
191
26
Sakashita
A
Hattori
T
Miller
CW
Suzushima
H
Asou
N
Takatsuki
K
Koeffler
HP
Mutation of p53 gene in adult T-cell leukemia.
Blood
79
1992
477
27
Nagai
H
Kinoshita
T
Imamura
J
Murakami
Y
Hayashi
K
Mukai
K
Ikeda
S
Tobinai
K
Saito
H
Shimoyama
M
Shimotohno
K
Genetic alteration of p53 in some patients with adult T-cell leukemia.
Jpn J Cancer Res
82
1991
1421
28
Yamato K, Oka T, Hiroi M, Iwahara Y, Sugito S, Tsuchida N, Miyoshi I: Aberrant expression of the p53 tumor suppressor gene in adult T-cell leukemia and HTLV-I-infected cells. Jpn J cancer Res 84:4, 1993
29
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 (ATL).
Blood
85
1995
2699
30
Yamada
Y
Hatta
Y
Murata
K
Sugawara
K
Ikeda
S
Mine
M
Maeda
T
Hirakata
Y
Kamihira
S
Tsukasaki
K
Ogawa
S
Hirai
H
Koeffler
HP
Tomonaga
M
Deletions of p15 and/or p16 genes as a poor-prognosis factor in adult T-cell leukemia.
J Clin Oncol
15
1997
1778
31
Hatta
Y
Yamada
Y
Tomonaga
M
Koeffler
HP
Extensive analysis of the retinoblastoma gene in adult T-cell leukemia/lymphoma (ATL).
Leukemia
11
1997
984
32
Uchida
T
Kinoshita
T
Watanabe
T
Nagai
H
Murate
T
Saito
H
Hotta
T
The CDKN2 gene alterations in various types of adult T-cell leukemia.
Br J Haematol
94
1996
665
33
Hatta
Y
Spirin
K
Tasaka
T
Morosetti
R
Said
JW
Yamada
Y
Tomonaga
M
Koeffler
HP
Analysis of p18INK4C in adult T-cell leukemia (ATL) and non-Hodgkin’s lymphoma.
Br J Haematol
99
1997
665
34
Shimoyama
M
Members of the Lymphoma Study Group (1984-87)
Diagnostic criteria and classification of clinical subtypes of adult T-cell leukemia-lymphoma.
Br J Haematol
79
1991
428
35
Hatta
Y
Takeuchi
S
Yokota
J
Koeffler
HP
Ovarian cancer has frequent loss of heterozygosity at chromosome 12p12.3-13.1 (region of TEL and Kip1 loci) and chromosome 12q23-ter: Evidence for two new tumor suppressor genes.
Br J Cancer
75
1997
1256
36
Hollstein
M
Sidransky
D
Vogelstein
B
Harris
CC
p53 mutations in human cancers.
Science
253
1991
49
37
Morita
R
Ishikawa
J
Tsutsumi
M
Hikiji
K
Tsukada
Y
Kamidono
S
Maeda
S
Nakamura
Y
Allelotype of renal cell carcinoma.
Cancer Res
51
1991
820
38
Devillee
P
van Vliet
M
van Sloun
P
Kuipers Dijkshoon
N
Hermans
J
Pearson
PL
Cornelisse
CJ
Allelotype of human breast carcinoma: A second major site for loss of heterozygosity is on chromosme 6q.
Oncogene
6
1991
1705
39
Lee
JH
Kavangah
JJ
Wildrick
DM
Wharton
JT
Block
M
Frequent loss of heterozygosity on chromosome 6q, 11, and 17 in human ovarican carcinoma.
Cancer Res
50
1990
2724
40
Saito
S
Saito
H
Koi
S
Sagae
S
Kudo
R
Saito
J
Noda
K
Nakamura
Y
Fine-scale deletion mapping of the distal long arm of chromosome 6 in 70 human ovarian cancers.
Cancer Res
52
1992
5815
41
De Souza
AT
Hankins
GR
Washington
MK
Fine
RL
Orton
TC
Jirtle
RL
Frequent loss of heterozygosity on 6q at the mannose 6-phosphate/insulin-like growth factor II receptor locus in human hepatocellular tumors.
Oncogene
10
1995
1725
42
Visakorpi
T
Kallioniemi
AH
Syvänen
A-C
hyytinen
ER
Karhu
R
Tammela
T
Isola
JJ
Kallioniemi
O-P
Genetic changes in primary recurrent prostate cancer by comparative genomic hybridization.
Cancer Res
55
1995
342
43
Trent
JM
Thompson
FH
Meyskens FL Jr
Identification of a recurring translocation site involving chromosome 6 in human malignant melanoma.
Cancer Res
49
1989
420
44
Millikin
D
Meese
E
Vogelstein
B
Witkowski
C
Trent
J
Loss of heterozygosity for loci on the arm of chromosome 6 in human malignant melanoma.
Cacner Res
51
1991
5449
45
Ray
ME
Su
YA
Meltzer
PS
Trent
J
Isolation and characterization of genes associated with chromosome-6 mediated tumor suppression in human malignant melanoma.
Oncogene
12
1996
2527
46
Healy
E
Belgaid
CE
Takata
M
Vahlquist
A
Rehman
I
Rigby
H
Rees
JL
Allelotypes of primary cutaneous melanoma and benign melanocytic nevi.
Cancer Res
56
1996
589
47
Gaidano
G
Hauptschein
RS
Parsa
NZ
Offit
K
Rao
PH
Lenoir
G
Knowles
DM
Chaganti
RSK
Dalla-Favera
R
Deletions involving two distinct regions of 6q in B-cell non-Hodgikin lymphoma.
Blood
80
1992
1781
48
Offit
K
Parsa
NZ
Gaidano
G
Filippa
DA
Louie
D
Pan
D
Jhanwar
SC
Dalla-Favera
R
Chaganti
RSK
6q deletions define distinct clinico-pathologic subsets of non-Hodgikin’s lymphoma.
Blood
82
1993
2157
49
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 leukemia.
Genes Chromosom Cancer
10
1994
286
50
Li
H
Lahti
JM
Valentine
M
Saito
M
Reed
SI
Look
AT
Kidd
VJ
Molecular cloning and chromosomal localization of the human cyclin C (CCNC) and cyclin E (CCNE) genes: Deletion of the CCNC gene in human tumors.
Genomics
32
1996
253
51
Guan
X-Y
Horsman
D
Zhang
HE
Parsa
NZ
Meltzer
PS
Trent
JM
Localization by chromosome microdissection of a recurrent breakpoint region on chromosome 6 in human B-cell lymphoma.
Blood
88
1996
1418
52
Gerard
B
Cave
H
Guidal
C
Dastugue
N
Vilmer
E
Grandchamp
B
Deletion of a 6cM commomly deleted region in childhood acute lymphoblastic leukemia on the 6q chromosomal arm.
Leukemia
11
1997
282
53
Tahara
H
Smith
AP
Gaz
RD
Cryns
VL
Arnold
A
Genomic localization of novel candidate tumor suppressor gene loci in human parathyroid adenomas.
Cancer Res
56
1996
599
54
Kamb
A
Gruis
NA
Weaver-Feldhaus
J
Liu
Q
Harshman
K
Tavtigian
SV
Stockert
E
Day
RS
Johnson
BE
Skolnick
MH
A cell cycle regulator potentially involved in genesis of many tumor types.
Science
264
1994
436
55
Nobori
T
Miura
K
Wu
DJ
Lois
A
Takabayashi
K
Carson
DA
Deletion of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers.
Nature
368
1994
753
56
Sadamori
N
Cytogenetic implication in adult T-cell leukemia: A hypothesis of leukemogenesis.
Cancer Genet Cytogenet
51
1991
131
57
Yamaguchi
T
Toguchida
J
Yamamuro
T
Kotoura
N
Takada
N
Kawaguchi
N
Kaneko
Y
Nakamura
Y
Sasaki
M
Ishizaki
K
Allelotype analysis in ostosarcomas: Frequent loss on 3q, 13q, 17p, and 18q.
Cancer Res
52
1992
2419
58
Tsuchiya
E
Nakamura
Y
Weng
S-Y
Nakagawa
K
Tsuchiya
S
Sugano
H
Kitagawa
T
Allelotype of non-small cell lung carcinoma: Comparison between loss of heterozygosity in squamous cell carcinoma and adenocarcinoma.
Cancer Res
52
1992
2478
Sign in via your Institution